425
Views
82
CrossRef citations to date
0
Altmetric
Review

Nanobody-derived nanobiotechnology tool kits for diverse biomedical and biotechnology applications

, , , , , , , & show all
Pages 3287-3303 | Published online: 21 Jul 2016

Abstract

Owing to peculiar properties of nanobody, including nanoscale size, robust structure, stable and soluble behaviors in aqueous solution, reversible refolding, high affinity and specificity for only one cognate target, superior cryptic cleft accessibility, and deep tissue penetration, as well as a sustainable source, it has been an ideal research tool for the development of sophisticated nanobiotechnologies. Currently, the nanobody has been evolved into versatile research and application tool kits for diverse biomedical and biotechnology applications. Various nanobody-derived formats, including the nanobody itself, the radionuclide or fluorescent-labeled nanobodies, nanobody homo- or heteromultimers, nanobody-coated nanoparticles, and nanobody-displayed bacteriophages, have been successfully demonstrated as powerful nanobiotechnological tool kits for basic biomedical research, targeting drug delivery and therapy, disease diagnosis, bioimaging, and agricultural and plant protection. These applications indicate a special advantage of these nanobody-derived technologies, already surpassing the “me-too” products of other equivalent binders, such as the full-length antibodies, single-chain variable fragments, antigen-binding fragments, targeting peptides, and DNA-based aptamers. In this review, we summarize the current state of the art in nanobody research, focusing on the nanobody structural features, nanobody production approach, nanobody-derived nanobiotechnology tool kits, and the potentially diverse applications in biomedicine and biotechnology. The future trends, challenges, and limitations of the nanobody-derived nanobiotechnology tool kits are also discussed.

Introduction

Over the decades, single-domain antibodies (sdAbs) have received a progressive interest from pharmaceutical and biotechnology industries owing to their peculiar properties, including small size, robust structure, high affinity and specificity, superior cryptic cleft accessibility, and deep tissue penetration.Citation1Citation3 The sdAbs could be categorized into man-made sdAbs and naturally occurring counterparts,Citation4 the latter including variable domain of heavy-chain-only antibodies in camelids (VHH), a naturally occurring antigen-binding variable domain of heavy chain from heavy-chain-only antibodies (HCAbs),Citation1 and variable domain of immunoglobulin new antigen receptors in sharks (V-NAR), a variable domain from immunoglobulin new antigen receptors (IgNARs).Citation5 Totally different from the man-made sdAbs, which have been pursued for the ideal ones by scientists for half a century, several naturally occurring sdAbs, including VHHs and V-NARs, already exist in nature in camelids or cartilaginous fish, sharing a surprising structural convergent evolution and performing similar biological functions.Citation1Citation5

In addition to the classical antibodies containing two heavy and two light chains, all camelidae including camels (Camelus dromedarius and Camelus bactrianus), llama (Lama glama and Lama guanicoe), and vicugna (Vicugna vicugna and Vicugna pacos) have HCAbs in their sera, lacking L chains and devoid of a canonical constant heavy-chain (CH) 1 domain.Citation1 Some cartilaginous fish, including nurse shark (Ginglymostoma cirratum), wobbegong (Orectolobus maculates), and dogfish (Squalus acanthias and Mustelus canis) sharks, also remarkably produce functional heavy-chain-only immunoglobulins (Igs), named IgNARs.Citation1,Citation5Citation8 Interestingly, some pathological and nonfunctional HCAbs were also discovered in sera of humans or in mouse hybridoma due to a partly genetic deletion of the variable heavy-chain (VH) and CH1 regions.Citation1 In camels or sharks, these HCAbs (or IgNARs) recognize the antigens via single variable domains, referred to as VHHs or V-NARs, respectively. Thus, the VHHs or V-NARs are the smallest intact antigen-binding domain derived from the HCAbs or IgNARs naturally occurring in camelids or cartilaginous fish.

Nanobody is referred to the VHHs, the sdAb fragments derived from naturally occurring camelid HCAbs or the counterpart domain V-NAR of IgNARs, the homodimeric antibodies devoid of light chains in sharks. Owing to the small dimensional size of 2.5 nm in diameter and 4 nm in height (~12–15 kDa), Ablynx in 2003 dubbed these sdAbs “nanobody” to emphasize their smaller dimensional sizes, compared to larger molecular sizes of single-chain variable fragments (scFvs; 27 kDa), antigen-binding fragments (Fabs; ~57kDa), and the intact conventional immunoglobulin-γ (IgG) antibody (~150 kDa).Citation3,Citation5,Citation8 In this review, we summarize the current state of the art in nanobody research, focusing on the nanobody structural features, nanobody production approach, nanobody-derived nanobiotechnology tool kits, and the potential applications in biomedicine and biotechnology.

Structure of nanobodies

In comparison with the conventional IgG antibody assembled from two identical heavy-chain and two identical light-chain (heterotetrameric structure, ), camelid HCAbs are HCAbs (homodimeric structure, ), devoid of the light-chain polypeptide and the first constant domain (CH1) of heavy-chain polypeptide.Citation1Citation3 The absence of light chains in the whole antibody and lack of the CH1 domain in the heavy chain are two significant features of camelid HCAbs, which provide them with a more compact architecture and smaller dimensional size with a molecular weight of ~90 kDa rather than ~150 kDa for the canonical antibody IgG.Citation1,Citation3 Similarly, the Ig isotype IgNAR (novel antigen receptor) discovered in the shark bloodstream is also a homodimeric structure of two heavy-chain polypeptides, each comprising a single variable domain and five constant domains (homodimeric structure, ).Citation7

Figure 1 Schematic representations of intact antibodies, including canonical antibodies (IgG1) and heavy-chain antibodies in camels (HCAbs) and sharks (IgNARs), and intact antibody-derived fragments.

Notes: (A) Intact canonical antibodies (IgG1) comprising two light chains (VL and CL domains) and two heavy chains (comprising VH, CH1, hinge, and CH2 and CH3 domains). (B) Canonical antibody-derived fragments: Fabs, scFvs, and VH domains. (C) The structure and packing of VH domain from canonical antibodies IgG1. (D) Intact HCAbs in camelids, comprising homodimeric heavy chains (containing VHH and CH2 and CH3 domains), devoid of light chains in intact antibodies and lack of CH1 domains in heavy chains. (E) Camel HCAb-derived single-domain antibodies: VHH. (F) The structure and packing of VHH from camel HCAbs. (G) Intact IgNARs in sharks, comprising homodimeric heavy chains (containing V-NAR and C1–C5 domains), devoid of light chains in antibodies. (H) Shark IgNAR-derived single-domain antibodies: V-NAR. (I) The structure and packing of V-NAR from shark IgNARs.

Abbreviations: CDRs, complementarity-determining regions; CH, constant heavy chain; CL, constant light chain; Fabs, antigen-binding fragments; HCAbs, heavy-chain-only antibodies; HV, hypervariable; Ig, immunoglobulin; IgNARs, Ig new antigen receptors; scFvs, single-chain variable fragments; VH, variable heavy chain; VHH, variable domain of HCAbs in camelids; VL, variable light chain; V-NAR, variable domain of IgNARs in sharks.

Figure 1 Schematic representations of intact antibodies, including canonical antibodies (IgG1) and heavy-chain antibodies in camels (HCAbs) and sharks (IgNARs), and intact antibody-derived fragments.Notes: (A) Intact canonical antibodies (IgG1) comprising two light chains (VL and CL domains) and two heavy chains (comprising VH, CH1, hinge, and CH2 and CH3 domains). (B) Canonical antibody-derived fragments: Fabs, scFvs, and VH domains. (C) The structure and packing of VH domain from canonical antibodies IgG1. (D) Intact HCAbs in camelids, comprising homodimeric heavy chains (containing VHH and CH2 and CH3 domains), devoid of light chains in intact antibodies and lack of CH1 domains in heavy chains. (E) Camel HCAb-derived single-domain antibodies: VHH. (F) The structure and packing of VHH from camel HCAbs. (G) Intact IgNARs in sharks, comprising homodimeric heavy chains (containing V-NAR and C1–C5 domains), devoid of light chains in antibodies. (H) Shark IgNAR-derived single-domain antibodies: V-NAR. (I) The structure and packing of V-NAR from shark IgNARs.Abbreviations: CDRs, complementarity-determining regions; CH, constant heavy chain; CL, constant light chain; Fabs, antigen-binding fragments; HCAbs, heavy-chain-only antibodies; HV, hypervariable; Ig, immunoglobulin; IgNARs, Ig new antigen receptors; scFvs, single-chain variable fragments; VH, variable heavy chain; VHH, variable domain of HCAbs in camelids; VL, variable light chain; V-NAR, variable domain of IgNARs in sharks.

The percentage of HCAbs in the bloodstream of camelids varies greatly among species because of a variation in mutation rates.Citation3 It might reach relatively high level in camels, ranging from ~50% to ~80%, whereas it totals up to 10%–25% in South American camelid species.Citation1 Relative to the HCAbs in camelidae, the shark IgNARs are slightly at lower level, ranging from ~0.1 mg/mL to 1 mg/mL, counting for ~5% of the total Igs in the bloodstream.Citation5,Citation9 The HCAbs and IgNARs have shown very high hypermutation, apparently in response to antigens in the immune protection of the camelid and the shark.Citation1,Citation5,Citation9 Therefore, to some extent, the nanobodies (VHHs or V-NARs) with a molecular weight of ~12–15 kDa in the HCAbs or IgNARs are the structural and functional counterparts of the Fabs in the conventional IgGs ().Citation1,Citation3,Citation5,Citation8

Similar to the VH domain in conventional IgGs, the folded VHH in camel HCAbs comprises nine β-strands. These β-strands, organized in a four-stranded β-sheet and a five-stranded β-sheet, were connected by three hypervariable (HV) loops, also called complementarity-determining regions (CDRs), and by a conserved disulfide bond between Cys23 within framework region (FR) 1 and Cys94 within FR3.Citation1 The three CDRs located at the N-terminal end of the domain forms a continuous surface (paratope) in response to recognizing epitopes of antigens. This means that VHHs of the camel HCAbs and VH domains of the conventional IgGs share the similar structural architecture of the FRs and loops ().Citation1,Citation2,Citation10,Citation11 The alignment of the VHH amino acid sequences indicates that two major significant differences between both the VHH and VH domains exist within the FR2 and in the CDRs, especially in the CDR3.

The first notable difference between VHH and VH is within FR2.Citation1,Citation3 The highly conserved hydrophobic amino acids found within FR2 of the conventional VH region at positions (Val37, Gly44, Leu45, and Trp47) are replaced by hydrophilic and/or smaller amino acids, mostly Phe42, Glu49, Arg50, and Gly52, respectively.Citation1,Citation12Citation15 These hydrophobic amino acids normally participate in the interaction with chaperone proteins and the variable light chain (VL) domain during the assembly of heavy chains and light chains of the conventional antibodies.Citation1 Thus, reshaping this VL side of the domain deprives the heavy chains of their binding sites for any VL interaction, enhancing solubility and reducing aggregation of VHH in the absence of a VL domain.Citation1,Citation3

The second difference between VHH and VH is found in the HV loops. There is a broader structural repertoire of loops in VHH, especially an enlarged HV region in the H1 loop (CDR1) and an extended H3 loop (CDR3) (). These uniquely extended loops provide a sufficient antigen-interacting surface, large enough to 600–800 ÅCitation2 as offered by six loops from the VH–VL domain pair in the traditional IgGs.Citation1,Citation3 It has been established that the elongated CDR3 is capable of extending into cavities on antigens, such as the active site crevice of enzymes and ligand-binding cavity of receptors.Citation1,Citation3 However, an enlarged loop suggests broader flexibility, expectedly leading to be entropically counterproductive for binding. This problem is solved in camel VHHs by fastening the extended loop of CDR3 with an extra disulfide bond toward either the CDR1 loop () or the CDR2 loop or even the FR2.Citation1,Citation3,Citation16 These disulfide bonds might maximally optimize the binding surface topology and facilitate the orientation of the CDR3 toward the antigens.Citation3

As for the V-NARs, it shows homology to Vα domain of the T-cell receptor.Citation5,Citation17 Contrary to VHH, V-NAR only consists of seven β-strands instead of nine β-strands due to the truncation in FR2–CDR2 (). This means that V-NARs have only two CDRs, ie, CDR1 and CDR3 (). Similar to the camelid VHHs, the V-NAR has abnormally long CDR3 loops that considerably reflect the greatest diversity of the V-NAR in both sequence and length.Citation7 Furthermore, highly frequent somatic mutation is also found in CDR1, at the equivalent site of the deleted CDR2, where the shorten loop forms a belt-like structure at the opposite side of the CDR1 and CDR3, and in a loop that resembles HV4 in T-cell receptors.Citation5,Citation18 Similar to the camel VHHs, the shark V-NARs also evolved to generate equivalent disulfide bridges tethering the extended antigen-binding loops.Citation1,Citation19,Citation20

Although the sequences of VHHs and V-NARs are very diverse,Citation1 the camel VHHs shared the similar architecture with V-NARs. Overall, both show a rugby ball-shaped structure and a convex or protruding surface at the N-terminal end of the domains.Citation19,Citation21 This protruding surface increases the actual interaction surface of the paratopes, extremely facilitating insertion of nanobodies in cavities on the surface of the antigens or ligand-binding sites of receptors.Citation22 In limited cases, a flat paratope surfaceCitation23 and occasionally a cavity for the antihapten bindersCitation24,Citation25 were also observed owing to the long loops in most nanobody structures folding over the FR2 region in VHH.Citation1 These different shapes of the paratope surfaces demonstrated the extreme flexibility and great diversity of both the sdAbs.

Nanobody libraries

To fish out a desired nanobody with high stability and subnanomolar or picomolar affinity, preparation of nanobody libraries using different approaches was reasonably proposed.Citation1,Citation4,Citation7,Citation26,Citation27 The technologies used for the preparation of nanobody library do not significantly differ from the ones otherwise used for recovering the Fab and scFv libraries. First of all, retrieval of nanobodies from an immune library is a priority consideration because somatic maturation in lymphocytes of immunized Camelidae will give antibody libraries more specific and higher affinity to antigens of interest.Citation4 However, for each new antigen, we have to prepare a new immune library, which might unnecessarily spend more time and costs than other strategies proposed where else, such as large one-pot libraries without immunization of animals.Citation4 Thus, a suitable naive library using blood samples from nonimmunized animals or the semisynthetic and synthetic libraries are practically alternative choices. Despite the lack of somatic maturation, it is possible that the selection based on phage display using such one-pot large libraries (>109 clones per library) allowed to isolate such VHHs with high affinity in the subnanomolar or picomolar range that are suitable for the diverse biomedical applications.

Immune library

Preparation of the immune nanobody library first needs an immunization of camelidae, through which antigen-specific HCAbs are affinity matured. The immunization procedures are mostly involved in prime–boost strategy using various antigens of interest as immunogens. After a brief immunization, the nanobodies are generally readily obtained by cloning the V gene repertoire from peripheral blood lymphocytes and by screening through phage display or other biological carriers.Citation1,Citation26 The entire Fab of the HCAb comprises only one VHH, and it containŝ120 amino acid residues, encoded by a gene fragment of onlŷ360 bp. Thus, the VHH gene is easily cloned by polymerase chain reaction (PCR) in one single amplicon. As a result, small libraries (just ~106 individual clones per library) created by usingã50 mL of blood sample already represent the immune VHH repertoire of lymphocytes present in bloodstream of the immunized animals.Citation1

As for the amplification and cloning of an scFv, the VH and VL exons needed to be first individually PCR amplified, which probably results in scrambled pairs of the VH and VL domains due to their random assembly. In contrast to the scFv cloning, the PCR amplification of the VHH present in only one exon generally facilitated generation of intact and affinity-matured VHHs from peripheral B-lymphocytes. Thus, the unique specificity and high affinity of nanobodies from immune libraries are guaranteed. Kinetic kon and koff rate constants of the nanobodies retrieved from the immune libraries are routinely reported to be low nanomolar or even picomolar levels, ranging from 105 to 106 M−1 s−1 and 10−2–10−4 s−1, respectively.Citation1 Such affinity parameters are excellent properties for the most biomedical applications, including disease diagnosis, bioimaging, drug screening, and targeting therapy.

Naive library

When toxic or nonimmunogenic antigens are potentially used as immunogens, or immunization is not available for some other reasons, naive V repertoires could be employed to replace the immune VHH libraries. This approach allows us saving the time and costs related to preparing a new library for any new antigens of interest. Due to the lack of somatic maturation stimulated in vivo by immunization process, such libraries theoretically need to includê109 individual clones to allow the retrieval of high-affinity binders to a given antigen in general.Citation1 Practically, the theoretical diversity of a naive library increases with increasing of the number of lymphocytes initially collected. Thus, large volume of blood samples (>1 L) collected from different animals is a prerequisite for preserving the greatest genetic diversity. In addition, to avoid unnecessary diversity reduction during cloning of this type of library, all of the necessary steps should be performed with the highest care to reduce the material loss. As a practical alternative, the final library size could also be beneficially increased by mixing independently prepared collections to guarantee the diversity of the library.Citation4

Semisynthetic/synthetic library

Limited size and diversity is a key problem that affects even large size naive-based nanobody libraries.Citation27 Without an enormous germline diversity and the recombinatorial diversity from VH/VL pairing in traditional IgGs, affinity maturation in camelids or sharks relies to a larger extent on somatic hypermutation that precisely tunes the CDRs to recognize any given antigens.Citation27 Thus, in an effort to mimic such in vivo diversification to yield diverse enough libraries capable of generating nanobodies to any given antigen, another strategy, ie, semisynthetic/synthetic library, was proposed. The high-affinity nanobody could also be fished out using this type of library without immunization of animals. The strategy is trying to conserve the FRs surrounding the CDRs, which may be crucial in conserving the structural integrity of nanobodies, and to randomly diversify the sequences of the CDRs, especially of CDR3.Citation27

Based on naturally occurring VHH or V-NAR sequences, the semisynthetic or synthetic nanobody libraries could be created by introducing length and sequence variations in CDR3 using randomized CDR3 primers,Citation18 or error-prone PCR combined with splice-overlap extension PCR method.Citation7,Citation27 Using small blood samples (<10 mL), the complexity of diversity-enhanced semisynthetic or synthetic nanobody library is close to 109, whereas the complexity of 106 of naive library could only be reached by using the same volume of blood samples. At least >1 L of blood samples might be consumed in order to obtaiñ109 individual clones per library for the naive library.Citation4 This means that the semisynthetic or synthetic library had better CDR3 diversity and better utility than the naturally occurring naive VHH or V-NAR libraries without immunization of animal or collection of large volume of blood samples. Thus, this may be a promising path toward obtaining a limitless source of nanobodies against a variety of antigens without immunization of animals.

Independent of the strategies to construct libraries, the established libraries further need to be displayed on different biological carries, including phages, bacteria, yeasts, and ribosomes, to facilitate screening and panning for a given antigen-specific nanobody. Retrieval of nanobodies from the libraries by phage display or any other selection protocols described earlier, including bacterial display, yeast display, intracellular two-hybrid selection, and ribosome display, has been well documented.Citation1 By a variety of standard biopanning strategies, these libraries are preciously hidden treasure with great molecular diversity and could be utilized to fish out the nanobody binders with desired properties to various antigens of interest.

Nanobody-derived nanobiotechnology tool kits

Within the new vista of nanobiotechnological applications, different nanosized biotools, nanoscaled biomacromolecules, and engineered bacteriophages have been employed as promising approaches to meet the unmet needs of biomedicine and biotechnology development for human health. Owing to the desired properties of nanobody, including nanoscaled size, stable and soluble behavior in aqueous solution, reversible refolding, humanizable sequences, and specific and high affinity for only one cognate target, as well as a sustainable source, nanobody has been an ideal research tool for the development of sophisticated nanobiotechnologies. Currently, the nanobody has been evolved into versatile research and application tool kits for diverse nanobiotechnology applications.Citation1Citation3,Citation10,Citation11,Citation28 A variety of nanobody-derived formats, including the nanobody itself, the radionuclide or fluorescent dye-labeled nanobodies, fluorescent protein or chromogenic enzyme fusion nanobodies, bivalent nanobodies, self-assembly motif-mediated nanobody homo-or heteromultimers, nanobody-coated nanoparticles, and nanobody-displayed bacteriophages (), have been successfully demonstrated as powerful nanobiotechnological tool kits for diverse biomedical applications, including targeting drug delivery and therapy,Citation1,Citation3 disease diagnosis,Citation2,Citation3 bioimaging,Citation11,Citation29Citation31 and agricultural and plant protection.Citation32,Citation33 These applications indicate a special advantage of these nanobody-derived technologies, already surpassing the “me-too” products of other equivalent binders, such as the full-length antibodies, scFvs, Fabs, targeting peptides, and DNA-based aptamers.

Figure 2 Schematic of versatile nanobody-derived nanobiotechnological tool kits containing nanobody itself (A), radionuclide-labeled (B) or fluorescent dye-labeled nanobodies (C), fluorescent protein fusion nanobodies (D), chromogenic enzyme fusion nanobodies (E), bivalent nanobodies (F), self-assembly motif-mediated nanobody homo- or heteromultimers (G), nanobody-coated nanoparticles (H), and nanobody-displayed phages (I and J).

Note: These nanobiotools have been successfully applied to a variety of biomedical applications.Citation1Citation3,Citation10,Citation11,Citation28,Citation34Citation37

Abbreviations: NPs, nanoparticles; VHH, variable domain of heavy-chain-only antibodies in camelids; V-NAR, variable domain of immunoglobulin new antigen receptors in sharks.

Figure 2 Schematic of versatile nanobody-derived nanobiotechnological tool kits containing nanobody itself (A), radionuclide-labeled (B) or fluorescent dye-labeled nanobodies (C), fluorescent protein fusion nanobodies (D), chromogenic enzyme fusion nanobodies (E), bivalent nanobodies (F), self-assembly motif-mediated nanobody homo- or heteromultimers (G), nanobody-coated nanoparticles (H), and nanobody-displayed phages (I and J).Note: These nanobiotools have been successfully applied to a variety of biomedical applications.Citation1–Citation3,Citation10,Citation11,Citation28,Citation34–Citation37Abbreviations: NPs, nanoparticles; VHH, variable domain of heavy-chain-only antibodies in camelids; V-NAR, variable domain of immunoglobulin new antigen receptors in sharks.

Versatile applications of nanobody-derived nanobiotechnologies

There are various biomedical applications using the nanobody-derived nanobiotechnologies, which has been extensively covered recently elsewhere.Citation1Citation3,Citation10,Citation11,Citation28,Citation34Citation37 Here, we focus on a number of examples, wherein nanobodies provide special advantages over other equivalent binders. These applications demonstrated a promising future of the use of nanobodies in versatile environments, including basic research, bioimaging, clinical diagnosis, therapeutics, and agricultural and plant protection. Compared to the conventional sdAbs, these diverse applications indicate the versatile and novel properties of nanobodies as promising sdAbs.

Nanobodies as versatile research tools in biotechnology

With the versatile properties of the nanobodies, they have been developed into various research tools used in basic research, including affinity purification, immunoprecipitation, chaperone-assisted crystallization, protein degradation, gene activation or inactivation, protein–protein interaction, and many others ().

Table 1 The typical applications of nanobody-derived nanobiotechnologies in basic biomedical research

Owing to the intrinsic stability, monomeric nature, and easy directional immobilization to solid substrates, the nanobody is considered an ideal ligand for biomolecule purifications. Compared to full-length antibodies, nanobodies could yield a high-column regeneration capacity, produce an increased amount of paratopes per gram of support materials, and only need milder elution conditions.Citation1Citation3 For example, antihuman IgG VHH has been developed for IgG purification and depletion from blood, outperforming canonical protein A-based method.Citation38 Apart from concave epitopes on properly folded proteins, nanobodies could recognize small linear peptide sequences, which has been confirmed by isolating anti-EPEA VHHs. EPEA is a C-terminal tetra-amino-acid Glu–Pro–Glu–Ala sequence that can be cloned as a tag behind any protein,Citation39 facilitating a rapid and robust affinity purification of proteins. This linear peptide sequence recognition by nanobodies has also been demonstrated by isolating VHHs against another tetra-amino-acid sequence KDEL, a C-terminal signature tag of endoplasmic reticulum-resident protein.Citation40 This anti-KDEL VHH nanobody was shown to be an excellent tool to study differences in ER-resident protein expression by recognizing the KDEL sequence at the C-terminus of proteins, irrespective of the protein context.Citation40

The high affinity and unique specificity of nanobodies also make themselves excellent candidates for immunoprecipitation applications and for chromatin immunoprecipitation with DNA microarray (ChIP-on-chip) technology, facilitating uncovering new transcription factor-binding sites.Citation2,Citation41 Furthermore, by the combination of nanobody and magnetosome, a VHH-coated magnetosome approach was proposed for in vitro and in vivo immunoprecipitation by magnetical recruitment of antigen partners from cell extracts or within living bacteria.Citation42 As membranous organelles present in magnetotactic bacteria, magnetosomes contain magnetite particles enabling orientation of bacteria in a magnetic field.Citation2 By expressing red fluorescent protein (RFP)-binding nano-body with a magnetosome membrane protein MamC, VHH-coated magnetosomes were generated to efficiently recognize and bind their antigens in vitro by magnetically separating the VHH particle–antigen complexes from unbound protein contaminants. Such VHH particles could potentially also be used for intracellular recognition and magnetosome recruitment of RFP-tagged proteins and their interaction partners within living bacteria.

Nanobodies were also used as effective chaperones to assist crystallization process and structural determination of membrane proteins and large protein complexes for which it is difficult to determine these protein structures by X-ray crystallography. Since the convex or protruding surface at the N-terminal end of nanobodies could be targeting and recognizing clefts on the surface of antigens and these clefts usually coincide with active enzymatic sites or ligand-/receptor-binding cavities, the nanobodies are very suitable chaperones to assist crystallization and structural determination of these challenging targets.Citation43,Citation44 It has been demonstrated that, compared to the conventionally full antibodies, such chaperones could facilitate crystal formation by maintaining highly dynamic proteins in one of the particular protein conformations and stabilize intrinsic flexible regions or detergent-solubilized membrane proteins through preventing hydrophobic surfaces from contact with solvent to facilitate the effective crystal formation ().Citation45Citation47 For example, β2-adrenergic receptor crystal has been elucidated in the presence of active state-specific VHH, which could stabilize the instable active state of the receptor,Citation2,Citation44 and a VHH also plays its unique roles as a chaperone for the formation of β2-microglobulin fibril by stabilizing early fibril intermediates and preventing their self-oligomerization.Citation2,Citation48 The structural information on the disordered N-terminal prion protein region has also been elucidated by a VHH-inhibiting prion oligomerization, eventually contributing to the understanding of early prion formation.Citation2,Citation49 Similarly, crystallization process of components of bacterial type 2 secretion system demonstrated that the VHHs could substantially facilitate well-diffracting crystal formation by merely providing additional contact surface to the target proteins.Citation2,Citation50,Citation51

Figure 3 Some challenging structures elucidated using nanobodies as crystallization chaperones.

Notes: (A) Nanobody Nb80 (red), as a structural mimic of GαS, stabilizes the active-state conformation of β2-adrenoreceptor (green).Citation44,Citation47 (B) Ribbon representation of the full-length human prion protein (HuPrP, green) in complex with nanobody Nb484 (red).Citation49 (C) A periplasmic N-terminal domain of GspD (green or cyan) from the type 2 secretion system secretin in complex with nanobody Nb7 (red), forming a compact GspD:Nb7 heterotetramer.Citation46,Citation51 (D) Nanobody Nb11 (red)-aided structure determination of EpsI (cyan):EpsJ (green) pseudopilin heterodimer, a component of the bacterial type 2 secretion system in Vibrio vulnificus.Citation50 Protein Data Bank accession numbers are given in parentheses.

Figure 3 Some challenging structures elucidated using nanobodies as crystallization chaperones.Notes: (A) Nanobody Nb80 (red), as a structural mimic of GαS, stabilizes the active-state conformation of β2-adrenoreceptor (green).Citation44,Citation47 (B) Ribbon representation of the full-length human prion protein (HuPrP, green) in complex with nanobody Nb484 (red).Citation49 (C) A periplasmic N-terminal domain of GspD (green or cyan) from the type 2 secretion system secretin in complex with nanobody Nb7 (red), forming a compact GspD:Nb7 heterotetramer.Citation46,Citation51 (D) Nanobody Nb11 (red)-aided structure determination of EpsI (cyan):EpsJ (green) pseudopilin heterodimer, a component of the bacterial type 2 secretion system in Vibrio vulnificus.Citation50 Protein Data Bank accession numbers are given in parentheses.

By an elegantly experimental design, another example of the use of nanobody is to trigger the depletion of antigen via the ubiquitin pathway. Caussinus et alCitation52,Citation53 designed a deGradFP nanocomplex to induce the degradation of protein in vivo. A proof of concept study was conducted in fruitfly (Drosophila melanogaster) and zebrafish (Danio rerio). The anti-green fluorescent protein (GFP) nanobody was fused to the F-box domain that recruits the polyubiquitination machinery. Once a target was captured in the ubiquitinated nanobody-based nanocomplex, the proteasome-mediated degradation could be initiated.Citation1,Citation52 This is a new protein knockout tool potentially for more sophisticated applications in biomedicine and biotechnology. The resulting ubiquitinated nanobody-based nanocomplex could be effectively restricted in certain tissues, and the extent of protein degradation could be real-time monitored by just measuring GFP fluorescence.Citation2

Recently, some other elegant works also demonstrated a nanobody-based system using fluorescent proteins as scaffolds for cell-specific gene manipulation.Citation54 Two different nanobodies that could bind different regions of GFP were fused to a transcriptional activation domain or a DNA-binding domain, respectively. As a result, the GFP expression in GFP-expressing cells could specifically lead to the formation of a nanoscaled active transcription factor.Citation54 This approach could be utilized to conveniently induce cell-specific transgene expression or gene knockdown by RNAi in a vast collection of transgenic GFP cell lines.Citation2

Nanobodies as powerful bioimaging reagents

Owing to small dimensional size and high affinity of the nanobodies against various targets of interest, eg, intracellular signaling molecules and cancer biomarkers, the nanobodies and their derivative formats used as versatile nanotracers have been successfully employed in this postgenomic era ().Citation1 Using fluorescent protein fusion nanobodies or anti-GFP nanobodies, some elegantly fabricated nanobody-based tracers have been developed for bioimaging in living cells. As a proof of concept, intracellular expression of the genetic fusion of a fluorescent protein with a nanobody produces useful chromobodies or fluobodies to trace in vivo intracellular target in various cellular compartments in living cells,Citation55,Citation56 avoiding the need of genetic modification of target proteins with fluorescent tags. For example, an anti-GFP nanobody, termed GFP-binding protein (GBP), was fused to a monomeric RFP to generate a chromobody. The resulting GFP-based chromobody could specifically label the intracellular GFP fusion proteins localized in cytoplasm or nuclear.Citation55 Several of these nanobody-based tracers were also developed for high-content analysis, eg, the direct visualization of endogenous native proteins or infectious HIV virions in living cells.Citation57,Citation58 Furthermore, the GBP is also applied in super-resolution microscopy for the visualization of GFP fusion proteins. By coupling organic dyes to GFP-binding nanobodies, the nanobody-based tracker could recognize any GFP-tagged construct, enabling single-molecule localization with super-resolution imaging techniques.Citation1,Citation59 Similarly, a GBP-coated gold nanoparticle was also employed as a single-molecule tracer to monitor GFP-tagged membrane proteins and is even capable of tracking intracellular proteins in living cells by internalization.Citation2,Citation60

Table 2 The representative applications of chromobodies-based nanobiotechnologies for intracellular bioimaging

Recently, the nanobody-based tracer was applied to study in vivo protein–protein interactions.Citation2 By a sophisticated experimental design, the GBP was first fused to an anchoring protein that will localize the GBP at a particular subcellular compartment. The fluorescent protein GFP or RFP was then fused to the two proteins of interest. Once interaction of the two proteins occurs, both the proteins tethered together will lead to a strong GFP–RFP colocalization signal at the subcellular compartment defined by the GBP. In the nucleus and cytosol of human cells, it has been successfully demonstrated the effectiveness of this approach. By this way, peptide inhibitors of protein–protein interactions in these intracellular environments were analyzed and screened.Citation2,Citation61 More importantly, intracellularly expressed nanobodies themselves remaining soluble and specific antigen recognition activities could also be used to interfere with particular protein functions by competing with normal in situ protein–protein interactions. Thus, the identification of such bioactive nanobodies could offer an opportunity for target validation and lead molecule optimization to investigate difficult interactions or interactions considered undruggable.Citation1,Citation62,Citation63

More interestingly, versatile nanotraps have been developed, by which nanobodies were exploited to report particular conformational variants of a target and even to alter target translocation and localization in different organelles of living cells.Citation64Citation67 For example, nanobodies could modulate the conformation and spectral properties of GFP. The tamoxifen-induced translocation of human estrogen receptor from the cytoplasm to the nucleus could be monitored in a sensitive, high-throughput manner by nuclear expression of a GFP-enhancing GBP and then by monitoring the ratio of imaging.Citation2,Citation64 Promyelocytic leukemia protein originally dispersed throughout the nucleus, whereas GFP fusion promyelocytic leukemia protein is specifically redirected to the nuclear lamina by coexpressing GBP-lamin1 as a nuclear lamina anchor.Citation2,Citation67 Contrary to trapping target proteins in nuclear lamina, GBP was also demonstrated to block the action of nuclear GFP fusion proteins by trapping them in the cytoplasm of plant cells.Citation2,Citation68

Nanobodies for disease diagnosis

Apart from the use of nanobodies as versatile bioimaging tools in living cells, nanobodies have been used as valuable in vivo detection probes for cancer, infectious disease, atherosclerotic lesions, inflammatory response, and many other diseases in preclinical and clinical environments ().

Table 3 The representative applications of nanobody-derived nanobiotechnologies for disease diagnosis in vitro and in vivo

The nanobodies could be quickly eliminated from bloodstream due to their small sizes with a molecular weight of ~12–15 kDa that is well below the renal clearance cutoff of molecular weight of ~50 kDa.Citation1,Citation69 Thus, the small-size property of nanobodies facilitates their fast extravasation, good tumor penetration, and rapid renal clearance, which eventually lead to rapid and sensitive imaging of target tissue just within a few hours postinjection.Citation2,Citation29,Citation30 This is requirement exactly for a good in vivo imaging agent.Citation1 Radionuclide-labeled nanobodies have been successfully applied to noninvasively image in vivo tumors via positron emission tomography and single-photon emission computed tomography. These nanobody-derived nanoprobes demonstrated a high target specificity, high stability, good solubility, high tumor-to-background signal, fast clearance of excess tracer, and low immunogenicity.Citation2 For example, 99mTc-labeled VHHs could be used to recognize HER2, a cancer antigen for breast cancer diagnosis in a preclinical setting,Citation29 to distinguish moderate and high expression of epidermal growth factor receptor for improved prognosis of cancer therapy,Citation30 to detect the status of inflammatory responses by imaging dendritic cells,Citation31 and to target VCAM1, an antigen used to diagnose vulnerable atherosclerotic plaques.Citation70

Besides the promising results obtained by using 99mTc-VHH with half-life time (t1/2) of ~6 hours, some short-lived nuclides were also tested for more patient-friendly diagnoses. Rapid targeting to diseased tissue and fast blood clearance of unbound nanobodies make it possible to use short-lived nuclide-labeled formats for in vivo diagnosis, such as 68Ga and 18F with the t1/2 of 68 minutes and 110 minutes, respectively. This approach makes it possible to measure picomolar concentrations of nanoprobe within 1–3 hours postinjection by using positron emission tomography and single-photon emission computed tomography imaging techniques, thus resulting in a very low radiation burden for patients.Citation1 Using a mouse epidermoid carcinoma A431 xenograft model, the 68Ga-labeled nanobodies against an epidermal growth factor receptor were recently tested for their performances, yielding a relatively high ratio of tumor to blood after 3 hours postinjection.Citation1,Citation71

Owing to rapid renal clearance of nanobodies, the disadvantage of using nanobodies as in vivo probes is high accumulation of unbound nanobodies in kidneys soon after the probe injection, finally leading to a high radiation dose in kidneys and a complicated analysis of nearby tissues.Citation2,Citation29,Citation31,Citation70 A method has been further proposed to reduce the background level encountered with radiolabeled VHHs by preinjection of unlabeled bivalent VHHs to first occupy all extratumoral sites.Citation72 Alternatively, nonradioactive VHH-based probes with near-infrared fluorophores have also been fabricated to image in vivo tumors. These near-infrared fluorophore-labeled VHHs also showed faster imaging compared to approved monoclonal antibodies (McAbs) targeting the same antigens.Citation2,Citation73,Citation74

Like a coin has two sides, the small size of nanobodies, however, might affect antigen–probe interaction when they are coated in adsorptive substrates in an in vitro enzyme linked immunosorbent assay (ELISA) for disease biomarkers in blood or other biopsies. Compared to traditional IgGs, when these small-size nanobodies are coated on adsorptive substrates, the paratopes of nanobodies are very close to the vicinity of adsorbing surface, which might hinder antigen–probe interaction.Citation75 C-terminal peptide extension could improve the accessibility of coated VHHs in ELISA, eg, by fusion to peptide fragments, including a myc-His-tag, a llama long hinge region-His-tag,Citation76 and an Fc chain.Citation2,Citation75 Through this way, the engineered camel sdAbs were immobilized for sensing human prostate-specific antigen, which demonstrated that a higher probe density mediates enhanced detection sensitivity in a surface plasmon resonance-based detection system.Citation75,Citation77 This means that the use of nanobodies to generate sensitive and selective biosensors for in vitro disease diagnosis is highly feasible.Citation2,Citation75

More particularly, the nanobodies have also been developed to detect disease biomarkers in human biopsies by antibody-based slide and bead arrays. For an instance, a biotinylated VHH has been applied to streptavidin beads for sensitive biomarker detection in patient sera.Citation2,Citation78 As for pathogen diagnosis, traditionally McAb-derived antibodies have been used for decades. Although these McAb-derived formats recognize pathogen antigens with high sensitivity, they often lack the required specificity, leading to unsatisfactory performances. It has been demonstrated that the performances of nanobodies far exceed traditional McAbs. For example, Brucella and Yersinia infections in livestock have been successfully distinguished by a VHH rather than conventional antibodies.Citation79 Similarly, Taenia solium infection in pigs could be successfully detected by species-specific VHHs, whereas the existing genus-specific McAbs have failed to discriminate between T. solium and Taenia hydatigena infections.Citation80 Both species-specific and genus-specific VHHs have been developed for the diagnosis of Trypanosoma parasites without purification of antigens, leading to easy species typing of the prevailing parasites.Citation2,Citation81 Recently, a general nanobody-based agglutination reagent, consisting of a fusion protein between a red blood cell-specific VHH and a disease antigen of interest, has been developed for diagnosing a variety of diseases when different disease-specific antigens are available. This elegantly designed system could be used for HIV diagnosis when an HIV-1 p24 antigen was fused with red blood cell-specific VHH.Citation82 Thus, agglutination mediated by anti-p24 antibodies in patient serum and the added p24-VHH fusion protein could be observed if the anti-p24 antibodies are present in HIV-positive patient sera.Citation2

Nanobodies as targeting therapeutics

The development of nanobodies as targeting therapeutics is still in a very early stage. Some elegant works have demonstrated that the use of the bioactive nanobodies for antitoxin, anti-infection, anti-inflammation, or enzyme inhibition is a potentially feasible way for novel therapeutic development (). For example, nanobodies have been evaluated for passive immunization to treat envenomed victims, demonstrating extreme high-neutralization potency. By far, nanobodies for antiscorpion toxins, antibacterial toxins, and anti-snake venom are actively being investigated.Citation83Citation87 Owing to their small size and extended CDR3, nanobodies also showed special advantages as therapeutics for infectious disease, including the infection of viruses, bacteria, and parasites, over conventional antibodies that usually obstruct the access of hidden and essential epitopes on pathogens.Citation88Citation96 The added value of the nanobodies as targeting therapeutics stems from their capacity to distinguish the cognate target from closely related variants. Most of the small organic antagonists or even the conventional antibodies to a larger extent cannot reach such high specificity. Therefore, nanobodies could be used to specifically inhibit unwanted enzymatic activities related to different cellular signaling pathways. For example, ecto-ADP-ribosyl transferase ART2.2-specific VHH effectively blocked the enzymatic and cytotoxic activities of ART2.2 in lymphatic organs following intravenous injection. This blockade was highly specific, ie, only targeting blockade of enzyme ART2.2 but not the related enzymes ART1 or ART2.1, which eventually leads to develop new antidotes against ADP-ribosylating toxins.Citation97 In addition, several other nanobody-derived therapeutics are already in the pipeline. For instances, anti-IL6R and anti-tumor necrosis factor α nanobodies were developed to treat rheumatoid arthritis,Citation98Citation100 and an anti-RANKL nanobody was generated for bone loss disorder therapy.Citation1 Some therapeutic nanobodies, such as an anti-von Willebrand factor nanobody for the treatment of acute thrombotic thrombocytopenic purpura, have passed Phase I and Phase II clinical trials.Citation101 However, there is expectedly high competition from other therapeutics, including conventional McAbs and other antibody-derived small-size formats, such as scFvs and Fabs.Citation1

Table 4 The representative applications of nanobody-derived nanobiotechnologies for potential targeting disease therapy

On the other hand, the success of nanobodies as targeting therapeutics in clinic environments will also come from a more patient friendly administration, such as topical, oral, and inhalation or targeting delivery of cargoes to tissues that are difficult to access, and from obtaining improved biopharmaceutical parameters, including blood concentrations and prolonged residence time.Citation1,Citation102 To this end, nanobodies have been extensively used for targeting delivery of cargoes to different targeting antigens in in vivo organs to improve their biopharmaceutical parameters. For example, nanobodies have been assessed in their capacity of crossing blood–brain barrier and transcytosis across epithelia, which will potentially lead to develop nanobody-based brain-targeting therapeutics.Citation103Citation106 Moreover, chemical conjugation of nanobodies to branched gold nanoparticles also effectively produced antigen-targeting photothermal therapeutics upon light irradiation in a near biological window.Citation107

Nanobodies for agricultural protection and food analysis

Besides the promising results from enzyme inhibition exerted by nanobodies in mammals, nanobodies could also be used as enzyme inhibitors to modulate plant physiology and increase starch content in plant. For example, an inhibitory VHH against starch-branching enzyme A has been generated to interfere with the function of enzymes in potato.Citation108 Because of specific enzyme inhibition by an inhibitory VHH, the content of amylose in potato tuber increases upon targeting the VHH to plastids. An increase in amylose is even higher than that of the antisense controls, demonstrating the unique performance of the nanobodies as plant enzyme inhibitors. Furthermore, nanobodies could be used as nanocarriers for more efficient and specific delivery of existing chemicals to crops and weeds. To this respect, plant-specific VHHs were covalently coupled to engineered agrocapsules to improve the chemical retention time at target sites and reduce the chemical amounts needed.Citation109 The plant-specific VHHs have also been generated that could bind stomata, trichomes, grass leaves, and the surface area of potato.Citation32 Importantly, these plant-specific VHHs also showed good tolerance for harsh field conditions, such as temperature, variable pH, and salt concentrations. Besides the crops and weeds, nanobodies have also been applied for specific and targeting delivery of insecticides to insects.Citation33,Citation109 To pursue a high-specific VHH, the VHH phages are selected directly against living aphids and even whole-insect ELISA has been developed to characterize these insect-specific VHHs. Interestingly, nanobodies also been applied to food analysis because of highly extreme themostability. Several anticaffeine VHHs have been already generated for the quantification of caffeine in hot beverages. At 70°C, one of these VHHs could recognize caffeine and amazingly recover its binding functionality after an incubation step at 90°C,Citation110 demonstrating the excellent thermostability of these nanobodies.

Conclusion and perspectives

In this postgenomic era, searching for picomolar affinity and high specific binders against different targets of interest, including proteins, peptides, DNAs, RNAs, polysaccharides, and small molecules, are overwhelming challenges. Compared to conventional antibodies, nanobodies have following excellent properties: nanoscale size, robust structure, stable and soluble behavior in aqueous solution, reversible refolding, high affinity and specificity for only one cognate target, superior cryptic cleft accessibility, and deep tissue penetration, as well as a sustainable source. It is the amazingly excellent properties of nanobodies that could meet the unmet demand for screening these novel types of binders from renewable nanobody resources, making them attractive alternatives to conventional antibodies and their single-domain formats, such as Fab and scFv. The nanobody-derived binders have already been demonstrated to not only recognize only one cognate target of interest with high affinity but also differentiate different conformations of these targets with superior specificity. Therefore, these unique properties of nanobodies offer us opportunities to develop specific nanobiotechnological tool kits for various biomedical and biotechnological applications.

Current research on applying the nanobody-derived nanobiotechnological tool kits in affinity purification, immunoprecipitation, chaperone-assisted crystallization, protein degradation, gene activation or inactivation, protein–protein interactions, cellular bioimaging, in vivo and in vitro disease diagnosis, targeting therapeutics, agricultural and plant protection, and food analysis indicate a promising future of the use of these nanobodies and their derivative formats in biomedical and biotechnological fields. In the future, application of nanobodies as versatile molecules in different fields, including targeting therapy, targeting delivery, immunosensors, and in vivo imaging, nano-based research tools, is highly anticipated. Among these future trends, commercialization of nanobodies as a next-generation targeting therapeutics is high priority and technical challenge.Citation111,Citation112 Although some nanobody-derived products are in the pipeline, some have passed Phase I and Phase II trials,Citation1,Citation101 the competition from conventional McAbs or other antibody-derived small-size formats is huge. As compared to conventional McAbs, the small size of nanobodies not only confers their good tumor penetration but also enables them for a rapid renal clearance, indicating the challenge to balance improved systemic distribution with decreased plasma half-life in designing nanobody-based targeting therapeutics.Citation111 Therefore, for some intended uses, properties and pharmacokinetics should be tailored by linking nanobodies to albumin-binding moieties or by changing their hydrodynamic volume in various ways to reach a high blood concentration over a prolonged blood residence time,Citation1,Citation102 ie, obtaining long-circulating nanobody-based therapeutics. Furthermore, several other obstacles, such as immunogenicity and functionalization,Citation3 still have to be clinically addressed before the use of nanobodies in the clinic as targeting therapeutics become feasible.

Disclosure

The authors report no conflicts of interest in this work.

Acknowledgments

This work was supported by the National Natural Science Foundation of China (YW, grant no 31470218; LY, grant no 81572022). The authors also thank the anonymous reviewers for their insightful suggestions on improving the original article.

References

  • MuyldermansSNanobodies: natural single-domain antibodiesAnnu Rev Biochem20138277579723495938
  • De MeyerTMuyldermansSDepickerANanobody-based products as research and diagnostic toolsTrends Biotechnol201432526327024698358
  • SiontorouCGNanobodies as novel agents for disease diagnosis and therapyInt J Nanomedicine201384215422724204148
  • SaerensDMuyldermansSSingle Domain Antibodies: Methods and ProtocolsTotowa, NJHumana Press2012
  • ZielonkaSEmptingMGrzeschikJKonningDBarelleCJKolmarHStructural insights and biomedical potential of IgNAR scaffolds from sharksMAbs201571152525523873
  • FlajnikMFKasaharaMOrigin and evolution of the adaptive immune system: genetic events and selective pressuresNat Rev Genet2010111475919997068
  • ShaoCYSecombesCJPorterAJRapid isolation of IgNAR variable single-domain antibody fragments from a shark synthetic libraryMol Immunol200744465666516500706
  • LiuJLAndersonGPDelehantyJBBaumannRHayhurstAGoldmanERSelection of cholera toxin specific IgNAR single-domain antibodies from a naive shark libraryMol Immunol20074471775178317007931
  • DooleyHFlajnikMFAntibody repertoire development in cartilaginous fishDev Comp Immunol2006301–2435616146649
  • MuyldermansSBaralTNRetamozzoVCCamelid immunoglobulins and nanobody technologyVet Immunol Immunopathol20091281–317818319026455
  • HelmaJCardosoMCMuyldermansSLeonhardtHNanobodies and recombinant binders in cell biologyJ Cell Biol2015209563364426056137
  • MuyldermansSAtarhouchTSaldanhaJBarbosaJAHamersRSequence and structure of VH domain from naturally occurring camel heavy chain immunoglobulins lacking light chainsProtein Eng199479112911357831284
  • VuKBGhahroudiMAWynsLMuyldermansSComparison of llama VH sequences from conventional and heavy chain antibodiesMol Immunol19973416–17112111319566760
  • HarmsenMMRuulsRCNijmanIJNiewoldTAFrenkenLGJde GeusBLlama heavy-chain V regions consist of at least four distinct subfamilies revealing novel sequence featuresMol Immunol2000371057959011163394
  • MaassDRSepulvedaJPernthanerAShoemakerCBAlpaca (Lama pacos) as a convenient source of recombinant camelid heavy chain antibodies (VHHs)J Immunol Methods20073241–2132517568607
  • GovaertJPellisMDeschachtNDual beneficial effect of inter-loop disulfide bond for single domain antibody fragmentsJ Biol Chem201228731970197922128183
  • CriscitielloMFSaltisMFlajnikMFAn evolutionarily mobile antigen receptor variable region gene: doubly rearranging NAR-TcR genes in sharksProc Natl Acad Sci U S A2006103135036504116549799
  • LiuJLAndersonGPGoldmanERIsolation of anti-toxin single domain antibodies from a semi-synthetic spiny dogfish shark display libraryBMC Biotechnol20077788718021450
  • StanfieldRLDooleyHVerdinoPFlajnikMFWilsonIAMaturation of shark single-domain (IgNAR) antibodies: evidence for induced-fit bindingJ Mol Biol2007367235837217258766
  • StanfieldRLDooleyHFlajnikMFWilsonIACrystal structure of a shark single-domain antibody V region in complex with lysozymeScience200430556911770177315319492
  • DesmyterATransueTRGhahroudiMACrystal structure of a camel single-domain VH antibody fragment in complex with lysozymeNat Struct Biol1996398038118784355
  • De GenstESilenceKDecanniereKMolecular basis for the preferential cleft recognition by dromedary heavy-chain antibodiesProc Natl Acad Sci U S A2006103124586459116537393
  • DesmyterADecanniereKMuyldermansSWynsLAntigen specificity and high affinity binding provided by one single loop of a camel single-domain antibodyJ Biol Chem200127628262852629011342547
  • SpinelliSTegoniMFrenkenLvan VlietCCambillauCLateral recognition of a dye hapten by a llama VHH domainJ Mol Biol2001311112312911469862
  • SpinelliSFrenkenLGHermansPCamelid heavy-chain variable domains provide efficient combining sites to haptensBiochemistry20003961217122210684599
  • HolligerPHudsonPJEngineered antibody fragments and the rise of single domainsNat Biotechnol20052391126113616151406
  • GoldmanERAndersonGPLiuJLFacile generation of heat-stable antiviral and antitoxin single domain antibodies from a semisynthetic llama libraryAnal Chem200678248245825517165813
  • HuangLMuyldermansSSaerensDNanobodies(R): proficient tools in diagnosticsExpert Rev Mol Diagn201010677778520843201
  • VaneyckenIDevoogdtNVan GassenNPreclinical screening of anti-HER2 nanobodies for molecular imaging of breast cancerFASEB J20112572433244621478264
  • HuangLGainkamLOTCaveliersVSPECT imaging with Tc-99m-labeled EGFR-specific nanobody for in vivo monitoring of EGFR expressionMol Imaging Biol200810316717518297364
  • De GroeveKDeschachtNDe KoninckCNanobodies as tools for in vivo imaging of specific immune cell typesJ Nucl Med201051578278920395332
  • JongedijkEVerheesenPinventorAgroSavfe NV, assignee (Gent, BE)Specific delivery of agrochemicalsUS patent85980811232013
  • VerheesenPJongedijkEinventorsAgroSavfe NV, assignee (Gent, BE)Insect binding antibodiesUS patent201302242268292013
  • HarmsenMMDe HaardHJProperties, production, and applications of camelid single-domain antibody fragmentsAppl Microbiol Biotechnol2007771132217704915
  • WesolowskiJAlzogarayVReyeltJSingle domain antibodies: promising experimental and therapeutic tools in infection and immunityMed Microbiol Immunol2009198315717419529959
  • de MarcoABiotechnological applications of recombinant single-domain antibody fragmentsMicrob Cell Fact2011104421658216
  • DmitrievOYLutsenkoSMuyldermansSNanobodies as probes for protein dynamics in vitro and in cellsJ Biol Chem201629183767377526677230
  • KloosterRMaassenBTHStamJCImproved anti-IgG and HSA affinity ligands: clinical application of VHH antibody technologyJ Immunol Methods20073241–211217570391
  • De GenstEJGuilliamsTWellensJStructure and properties of a complex of alpha-synuclein and a single-domain camelid antibodyJ Mol Biol2010402232634320620148
  • KloosterREmanMRle DucQSelection and characterization of KDEL-specific VHH antibody fragments and their application in the study of ER resident protein expressionJ Immunol Methods20093421–211219041652
  • Nguyen-DucTPeetersEMuyldermansSCharlierDHassanzadeh-GhassabehGNanobody(R)-based chromatin immunoprecipitation/micro-array analysis for genome-wide identification of transcription factor DNA binding sitesNucleic Acids Res2013415e5923275538
  • PollithyARomerTLangCMagnetosome expression of functional camelid antibody fragments (nanobodies) in Magnetospirillum gryphiswaldenseAppl Environ Microbiol201177176165617121764974
  • BaranovaEFronzesRGarcia-PinoASbsB structure and lattice reconstruction unveil Ca2+ triggered S-layer assemblyNature2012487740511912222722836
  • RasmussenSGFChoiH-JFungJJStructure of a nanobody- stabilized active state of the beta(2) adrenoceptorNature2011469732917518021228869
  • LorisRMarianovskyILahJCrystal structure of the intrinsically flexible addiction antidote MazEJ Biol Chem200327830282522825712743116
  • KoideSEngineering of recombinant crystallization chaperonesCurr Opin Struct Biol200919444945719477632
  • PardonELaeremansTTriestSA general protocol for the generation of nanobodies for structural biologyNat Protoc20149367469324577359
  • DomanskaKVanderhaegenSSrinivasanVAtomic structure of a nanobody-trapped domain-swapped dimer of an amyloidogenic beta 2-microglobulin variantProc Natl Acad Sci U S A201110841314131921220305
  • AbskharonRNNGiachinGWohlkonigAProbing the N-terminal beta-sheet conversion in the crystal structure of the human prion protein bound to a nanobodyJACS20141363937944
  • LamAYPardonEKorotkovKVHolWGJSteyaertJNanobody-aided structure determination of the EpsI:EpsJ pseudopilin heterodimer from Vibrio vulnificusJ Struct Biol2009166181519118632
  • KorotkovKVPardonESteyaertJHolWGJCrystal structure of the N-terminal domain of the secretin GspD from ETEC determined with the assistance of a nanobodyStructure200917225526519217396
  • CaussinusEKancaOAffolterMFluorescent fusion protein knockout mediated by anti-GFP nanobodyNat Struct Mol Biol201119111712122157958
  • CaussinusEKancaOAffolterMProtein knockouts in living eukaryotes using deGradFP and green fluorescent protein fusion targetsCurr Protoc Protein Sci20137373 Unit30.2
  • TangJCYSzikraTKozorovitskiyYA nanobody-based system using fluorescent proteins as scaffolds for cell-specific gene manipulationCell2013154492893923953120
  • RothbauerUZolghadrKTillibSTargeting and tracing antigens in live cells with fluorescent nanobodiesNat Methods200631188788917060912
  • OlichonASurreyTSelection of genetically encoded fluorescent single domain antibodies engineered for efficient expression in Escherichia coliJ Biol Chem200728250363143632017921141
  • ZolghadrKGregorJLeonhardtHRothbauerUCase study on live cell apoptosis-assay using lamin-chromobody cell-lines for high-content analysisMethods Mol Biol201291156957522886277
  • HelmaJSchmidthalsKLuxVDirect and dynamic detection of HIV-1 in living cellsPLoS One2012711E5002623209635
  • RiesJKaplanCPlatonovaEEghlidiHEwersHA simple, versatile method for GFP-based super-resolution microscopy via nanobodiesNat Methods20129658258422543348
  • LeducCSiSGautierJA highly specific gold nanoprobe for live-cell single-molecule imagingNano Lett20131341489149423458263
  • HerceHDDengWHelmaJLeonhardtHCardosoMCVisualization and targeted disruption of protein interactions in living cellsNat Commun20134266024154492
  • TanakaTSewellHWatersSPhillipsSEVRabbittsTHSingle domain intracellular antibodies from diverse libraries: emphasizing dual functions of LMO2 protein interactions using a single VH domainJ Biol Chem201128653707371620980262
  • Perez-MartinezDTanakaTRabbittsTHIntracellular antibodies and cancer: new technologies offer therapeutic opportunitiesBioessays201032758959820544739
  • KirchhoferAHelmaJSchmidthalsKModulation of protein properties in living cells using nanobodiesNat Struct Mol Biol201017113313820010839
  • RomerTLeonhardtHRothbauerUEngineering antibodies and proteins for molecular in vivo imagingCurr Opin Biotechnol201122688288721708456
  • SchmidthalsKHelmaJZolghadrKRothbauerULeonhardtHNovel antibody derivatives for proteome and high-content analysisAnal Bioanal Chem201039783203320820372881
  • RothbauerUZolghadrKMuyldermansSSchepersACardosoMCLeonhardtHA versatile nanotrap for biochemical and functional studies with fluorescent fusion proteinsMol Cell Proteomics20087228228917951627
  • SchornackSFuchsRHuitemaERothbauerULipkaVKamounSProtein mislocalization in plant cells using a GFP-binding chromobodyPlant J200960474475419686537
  • VaneyckenID’HuyvetterMHernotSImmuno-imaging using nanobodiesCurr Opin Biotechnol201122687788121726996
  • BroisatAHernotSToczekJNanobodies targeting mouse/human VCAM1 for the nuclear imaging of atherosclerotic lesionsCirc Res2012110792793722461363
  • VosjanMJWDPerkLRRooversRCFacile labelling of an anti-epidermal growth factor receptor nanobody with Ga-68 via a novel bifunctional desferal chelate for immuno-PETEur J Nucl Med Mol Imaging201138475376321210114
  • MovahediKSchoonoogheSLaouiDNanobody-based targeting of the macrophage mannose receptor for effective in vivo imaging of tumor-associated macrophagesCancer Res201272164165417722719068
  • KijankaMWarndersF-JEl KhattabiMRapid optical imaging of human breast tumour xenografts using anti-HER2 VHHs site-directly conjugated to IRDye 800CW for image-guided surgeryEur J Nucl Med Mol Imaging201340111718172923778558
  • OliveiraSvan DongenGAMSStigter-van WalsumMRapid visualization of human tumor xenografts through optical imaging with a near-infrared fluorescent anti-epidermal growth factor receptor nanobodyMol Imaging2012111334622418026
  • SaerensDFrederixFReekmansGEngineering camel single-domain antibodies and immobilization chemistry for human prostate-specific antigen sensingAnal Chem200577237547755516316161
  • HarmsenMMFijtenHPDImproved functional immobilization of llama single-domain antibody fragments to polystyrene surfaces using small peptidesJ Immunoassay Immunochem201233323425122738648
  • SaerensDHuangLBonroyKMuyldermansSAntibody fragments as probe in biosensor developmentSensors20088846694686
  • Even-DesrumeauxKBatyDChamesPStrong and oriented immobilization of single domain antibodies from crude bacterial lysates for high-throughput compatible cost-effective antibody array generationMol Biosyst20106112241224820859568
  • AbbadyAQAl-DaoudeAAl-MaririAZarkawiMMuyldermansSChaperonin GroEL a Brucella immunodominant antigen identified using nanobody and MALDI-TOF-MS technologiesVet Immunol Immunopathol20121463–425426322472910
  • DeckersNSaerensDKanobanaKNanobodies, a promising tool for species-specific diagnosis of Taenia solium cysticercosisInt J Parasitol200939562563319041315
  • SaerensDStijlemansBBaralTNParallel selection of multiple anti-infectome nanobodies without access to purified antigensJ Immunol Methods20083291–213815017996887
  • HabibISmolarekDHattabCVHH (nanobody) directed against human glycophorin A: a tool for autologous red cell agglutination assaysAnal Biochem20134381828923541519
  • HmilaISaerensDBen AbderrazekRA bispecific nanobody to provide full protection against lethal scorpion envenomingFASEB J20102493479348920410443
  • HussackGArbabi-GhahroudiMvan FaassenHNeutralization of Clostridium difficile toxin A with single-domain antibodies targeting the cell receptor binding domainJ Biol Chem2011286118961897621216961
  • StewartCSMacKenzieCRHallJCIsolation, characterization and pentamerization of alpha-cobrotoxin specific single-domain antibodies from a naive phage display library: preliminary findings for antivenom developmentToxicon200749569970917257638
  • CookDANSamarasekaraCLWagstaffSCKinneJWerneryUHarrisonRAAnalysis of camelid IgG for antivenom development: immunoreactivity and preclinical neutralisation of venom-induced pathology by IgG subclasses, and the effect of heat treatmentToxicon201056459660320547172
  • HarrisonRAHassonSSHarmsenMLaingGDConrathKTheakstonRDGNeutralisation of venom-induced haemorrhage by IgG from camels and llamas immunised with viper venom and also by endogenous, non-IgG components in camelid seraToxicon200647336436816359717
  • PantNMarcotteHHermansPLactobacilli producing bispecific llama-derived anti-rotavirus proteins in vivo for rotavirus-induced diarrheaFuture Microbiol20116558359321585264
  • KrugerCHultbergAMarcotteHTherapeutic effect of llama derived VHH fragments against Streptococcus mutans on the development of dental cariesAppl Microbiol Biotechnol200672473273716636830
  • HarmsenMMFijtenHPDEngelBDekkerAEblePLPassive immunization with llama single-domain antibody fragments reduces foot-and-mouth disease transmission between pigsVaccine200927131904191119368770
  • StrokappeNSzynolAAasa-ChapmanMLlama antibody fragments recognizing various epitopes of the CD4bs neutralize a broad range of HIV-1 subtypes A, B and CPLoS One201273e3329822438910
  • KohWWLSteffensenSGonzalez-PajueloMGeneration of a family-specific phage library of llama single chain antibody fragments that neutralize HIV-1J Biol Chem201028525191161912420400507
  • HinzAHulsikDLForsmanACrystal structure of the neutralizing llama V-HH D7 and its mode of HIV-1 gp120 interactionPLoS One201055e1048220463957
  • De HaardHJWBezemerSLedeboerAMLlama antibodies against a lactococcal protein located at the tip of the phage tail prevent phage infectionJ Bacteriol2005187134531454115968064
  • IbanezLIDe FiletteMHultbergANanobodies with in vitro neutralizing activity protect mice against H5N1 influenza virus infectionJ Infect Dis201120381063107221450996
  • StijlemansBConrathKCortez-RetamozoVEfficient targeting of conserved cryptic epitopes of infectious agents by single domain antibodies – African trypanosomes as paradigmJ Biol Chem200427921256126114527957
  • Koch-NolteFReyeltJSchossowBSingle domain antibodies from llama effectively and specifically block T cell ecto-ADP-ribosyltransferase ART2.2 in vivoFASEB J200721133490349817575259
  • Camacho-VillegasTMata-GonzalezTPaniagua-SolisJSanchezELiceaAHuman TNF cytokine neutralization with a vNAR from Heterodontus francisci shark: a potential therapeutic useMAbs201351808523221782
  • BojalilRMata-GonzalezMTSanchez-MunozFAnti-tumor necrosis factor (V)NAR single domains reduce lethality and regulate underlying inflammatory response in a murine model of endotoxic shockBMC Immunol2013141723548047
  • Van RoyMVerverkenCBeirnaertEThe preclinical pharmacology of the high affinity anti-IL-6R Nanobody® ALX-0061 supports its clinical development in rheumatoid arthritisArthritis Res Ther20151713525994180
  • HolzJ-BThe TITAN trial – assessing the efficacy and safety of an anti-von Willebrand factor nanobody in patients with acquired thrombotic thrombocytopenic purpuraTrans Apher Sci2012463343346
  • KontermannREStrategies for extended serum half-life of protein therapeuticsCurr Opin Biotechnol201122686887621862310
  • LiTBourgeoisJ-PCelliSCell-penetrating anti-GFAP VHH and corresponding fluorescent fusion protein VHH-GFP spontaneously cross the blood-brain barrier and specifically recognize astrocytes: application to brain imagingFASEB J201226103969397922730440
  • PerruchiniCPecorariFBourgeoisJ-PDuyckaertsCRougeonFLafayePLlama VHH antibody fragments against GFAP: better diffusion in fixed tissues than classical monoclonal antibodiesActa Neuropathol2009118568569519597828
  • IqbalUTrojahnUAlbaghdadiHKinetic analysis of novel mono-and multivalent VHH-fragments and their application for molecular imaging of brain tumoursBr J Pharmacol201016041016102820590596
  • EmmersonCDvan der VlistEJBraamMREnhancement of polymeric immunoglobulin receptor transcytosis by biparatopic VHHPLoS One2011610e2629922022593
  • Van de BroekBDevoogdtND’HollanderASpecific cell targeting with nanobody conjugated branched gold nanoparticles for photothermal therapyACS Nano2011564319432821609027
  • JoblingSAJarmanCTehMMHolmbergNBlakeCVerhoeyenMEImmunomodulation of enzyme function in plants by single-domain antibody fragmentsNat Biotechnol2003211778012483224
  • VerheesenPDe JongheCinventorAgroSavfe NV, assignee (Gent, BE)Manufacturing of specifically targeting microcapsulesUS patent201500875173262015
  • LadensonRCCrimminsDLLandtYLadensonJHIsolation and characterization of a thermally stable recombinant anti-caffeine heavy-chain antibody fragmentAnal Chem200678134501450816808459
  • TurnerKBAlvesNJMedintzILWalperSAImproving the targeting of therapeutics with single-domain antibodiesExpert Opin Drug Del2016134561570
  • KijankaMDorresteijnBOliveiraSHenegouwenPNanobody-based cancer therapy of solid tumorsNanomedicine (Lond)201510116117425597775